• Aucun résultat trouvé

The PAX5 oncogene is expressed in N-type neuroblastoma cells and increases tumorigenicity of a S-type cell line

N/A
N/A
Protected

Academic year: 2021

Partager "The PAX5 oncogene is expressed in N-type neuroblastoma cells and increases tumorigenicity of a S-type cell line"

Copied!
8
0
0

Texte intégral

(1)

The PAX5 oncogene is expressed in N-type neuroblastoma cells and increases

tumorigenicity of a S-type cell line

Florian B.Baumann Kubetzko

1

, Claudio di Paolo

1

,

Charlotte Maag

1

, Roland Meier

3

, Beat W.Schafer

4

,

David R.Betts

4

, Rolf A.Stahel

1

and Andreas

Himmelmann

1,2,5

1Forschungslabor Molekulare Onkologie, Klinik und Poliklinik fur Onkologie, Universitaetsspital Zurich, Haeldeliweg 4, CH-8044 Zurich, Switzerland,2Medizinische Klinik B, Universitaetsspital Zurich, Raemistrasse 100, CH-8091 Zurich, Switzerland,3Medizinische Klinik, Universitaets-Kinderklinik, Steinwiesstrasse 75, CH-8032 Zurich, Switzerland and 4Abteilung fur Onkologie, Universitaets-Kinderklinik, Steinwiesstrasse 75, CH-8032 Zurich, Switzerland

5To whom correspondence should be addressed Email: andreas.himmelmann@usz.ch

Neuroblastoma is a neural crest-derived neoplasm of

infancy with poor outcome in patients with advanced

disease. The oncogenic transcription factor PAX5 is an

important developmental regulator and is implicated in the

pathogenesis of several malignancies. Screening of

neuro-blastoma cell lines revealed PAX5 expression in a

malig-nant subset of neuroblastoma cells, so-called `N-type' cells,

but not in the more benign `S-type' neuroblastoma cells.

PAX5 expression was also detected in small cell lung

cancer, an aggressive tumor of neural crest origin. Based

on this observation we hypothesized that there could be a

relationship between PAX5 expression and the more

malig-nant phenotype of N-type cells. Stable PAX5 expression

was established in several clones of the S-type cell line

CA-2E. A noticeable difference in morphology of these

transfectants was observed and there was also a significant

increase in the proliferation rate. Moreover, PAX5

expressing clones gained the ability to form colonies in a soft

agar assay, a marker of tumorigenicity. Down-regulation

of PAX5 in several N-type cell lines and one small cell lung

cancer cell line utilizing small interfering RNA resulted in

a significant decrease in growth rate. Taken together we

propose PAX5 as an important factor for the maintenance

of the proliferative and tumorigenic phenotype of

neuro-blastoma. Our data, together with a recent study on the

role of PAX genes in cancer suggest that PAX5 and other

PAX transcription factors might be valuable targets for

cancer therapy.

Introduction

Neuroblastoma is the most common extracranial solid tumor in

children and is the leading cause of cancer death in children

between 1 and 4 years (1,2). It is a neoplasm of the neural crest,

a transient embryonic structure giving rise to a variety of

tissues (e.g. melanocytes, neurons, neuroendocrine tissue and

smooth muscle cells) (3). Histologically neuroblastoma tumors

are composed of distinct malignant cell types, including

neu-roblastic and stromal cells (1).

Cellular diversity is also characteristic of cell lines

estab-lished from explanted neuroblastoma tumors. A number of

sub-populations have been identified in these cultures, including

neuroblastic (N-type), substrate adherent (S-type) cells and

cells with an intermediate phenotype (I-type). These cell

types can be distinguished by a characteristic morphology

(4--6). Analysis of homogeneous cultures of these subtypes

has shown that they can also be distinguished by the expression

of a number of differentiation-associated markers. N-type cells

express neurotransmitter biosynthetic enzymes, chromogranin

A and neurofilament proteins while S-type cells synthesize

collagen, fibronectin and vimentin (4,7). In contrast to S-type

cells, N-type cells are tumorigenic in nude mice and form

colonies in soft agar (8). The difference in malignant

proper-ties between S- and N-type cell subtypes may have clinical

relevance (9). At the transcriptional level several differences in

gene expression have been detected. For example, BCL2 and

MYCN are usually expressed at a higher level in N-type cells

(10,11). The difference in MYCN expression was observed in

both MYCN amplified and non-amplified cell lines (11,12).

Recently it has been demonstrated that S-type in contrast to

N-type cells are more susceptible to TRAIL-mediated

apopto-sis due to their expression of caspase-8 (13,14).

PAX5 is a member of the PAX gene family of

developmen-tally regulated transcription factors that play a fundamental

role in organogenesis (15). All PAX transcription factors share

a DNA binding domainÐthe paired box. Their importance in

development has been underscored by several loss-of-function

mutations that cause lack of a specific structure or organ in

which the corresponding PAX gene is normally expressed.

Typically, PAX protein expression is down-regulated in the

adult organism. During mouse brain development Pax5 is

expressed predominantly at the midbrain--hindbrain boundary.

A weaker expression is found along the entire neural tube,

particularly in the ventricular zone of both alar and basal plates

(16). A similar expression pattern has been described recently

in human embryos (17). In addition, Pax5 is expressed during

all stages of B-cell development, except in plasma cells (18).

Pax5

ÿ=ÿ

mice lack mature B cells and show defects of the

inferior colliculus and anterior cerebellum (19).

In addition to the normal physiological function during

development, PAX transcription factors have been shown to

play an important role in tumorigenesis (reviewed in ref. 20).

First, a number of PAX genes are located at recurring,

tumor-specific chromosomal translocations, suggesting that PAX

genes have an oncogenic capacity when inappropriately

expressed. Secondly, several PAX genes are re-expressed in

malignant neoplasms, usually in tumors derived from tissue in

which the respective PAX gene is expressed during

develop-ment. For example, deregulated expression of PAX5 has been

described in medulloblastoma and lymphomas (21,22) and of

Abbreviations: MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide; NSCLC, non-small cell lung cancer; SCLC, small cell lung cancer; scRNA, scrambled control; siRNA, small interfering RNA.

(2)

PAX3 in melanoma (23). It is speculated that the re-expressed

PAX genes promote tumor development and progression

by increasing proliferation and motility, while inhibiting

apoptosis (20).

Here we describe that PAX5 is expressed in aggressive

N-type neuroblastoma cell lines, while no expression was

detected in S-type cells. Over-expression of PAX5 in the

S-type cell line CA-2E restored several malignant properties,

in particular anchorage-independent growth. In addition

down-regulation of PAX5 in several N-type cell lines significantly

reduced their proliferation rate. These results provide

addi-tional evidence for an important role of the PAX gene family

in tumor development and suggest a role for PAX5 in

neuro-blastoma disease progression.

Materials and methods

Cell lines

ACN, CA-2E, BE(2)C, BE(2)M17, Imr32, Kelly, LAN-1, LAN-5, SH-310, SH-EP, SH-IN, SK-N-BE(2), SH-SY5Y and WSN were kindly provided by Nicole Gross (University Hospital, Lausanne, Switzerland). U1285 was a kind gift of Larisa Belyanskaya (Universitaetsspital, Zurich, Switzerland), MMLI157 was provided by Juerg Zarn (formerly Universitaetsspital Zurich, Switzerland) and Kis-1 was kindly provided by Hitoshi Ohno (Kyoto Univer-sity, Kyoto, Japan). SW2, OH1 and OH3 have been described previously (24). A549, Calu1, Calu3, Calu6, H460, NCI-H69 and NCI-H125 were obtained from ATCC (Rockville, MD). All cell lines were cultured in RPMI1640 supplemented with 10% FCS, 2 mML-glutamine, 50 IU/ml penicillin and 50 mg/ml streptomycin and maintained at 37C in a humified atmosphere with 5% CO2.

RT--PCR

RNA was isolated from cell lines using the RNeasy Mini Kit according to the manufacturer's instructions (Qiagen, Basel, Switzerland). Five micro-grams of total RNA were used to synthesize cDNA using the M-MuLV reverse transcriptase according to the manufacurer's instructions (MBI Fermentas, Vilnius, Lithuania). One-hundredth of the product was used for amplification using primers for PAX5 or GAPDH and PCR conditions as described elsewhere (25).

Plasmid construction

A mammalian PAX5 cDNA expression plasmid designated pX-13 was gener-ated by cloning a 3.3 kb XbaI fragment from phBSAP-1s (a kind gift of Meinrad Busslinger, Research Institute of Molecular Pathology, Vienna, Austria) into the mammalian expression vector pcDNA3.1/Hygro(‡) (Invitrogen, Basel, Switzerland) using standard methods (26). This fragment contained the full-length PAX5 coding sequence (1176 bp) together with 77 bp of the 50-UTR and 2026 bp of the 30-UTR sequence. The correct orientation of the PAX5-cDNA was confirmed by sequencing (Microsynth, Balgach, Switzerland).

Cell transfection and generation of stable cell lines

Transfection of the plasmids was performed with Superfect following the manufacturer's instructions (Qiagen, Basel, Switzerland). Stable clones were generated by transfecting CA-2E cells with plasmids pX-13 or pcDNA3.1/ Hygro(‡) for 20 h. The transfection medium was aspirated and cells were cul-tured in fresh medium for another day before adding 400 mg/ml hygromycin B for selection. Single clones were achieved by plating the cells in 96-well plates after 7--10 days of selection at a density of 0.5 cells/well in 100 ml selective medium. Single clones were expanded and screened for PAX5 expression by immunoblotting.

Cell lysis and immunoblotting

Cells were harvested and pellets were frozen at ÿ80C for at least 1 h. RIPA buffer was used for cell lysis as described (22). Between 80 and 120 mg of total lysate was separated by SDS--PAGE on a 10% gel. Transfer and immunoblotting conditions have been described previously (24). For PAX5 and actin detection the Pax5-C14 antibody (Santa Cruz Biotechnology, Santa Cruz, CA) or an anti-Actin antibody (ICN Biomedicals GmbH, Eschwege, Germany) have been used respectively and were combined with the corre-sponding secondary antibodies. Chemoluminescent signals were detected using the ECL Kit (Amersham Biosciences, Buckinghamshire, UK). Band intensities were measured using standard methods.

Growth experiments

5000 cells were plated in 6-well plates containing 3 ml RPMI 1640 supple-mented with 5% FCS, 2 mML-glutamine, 50 IU/ml penicillin and 50 mg/ml streptomycin. Cells were grown at standard conditions and harvested after 4 days. After centrifugation the cells were re-suspended in PBS and the number of cells was determined using a Coulter counter (Beckman Coulter, Fullerton, CA). The harvesting and counting procedure was repeated every 24 h for an additional 5 days. Experiments were performed in triplicate and repeated four times with similar results.

Soft agar assay

To determine the ability of anchorage-independent growth a soft agar clono-genic assay was performed with PAX5 positive and negative clones. A base solution consisting of 1.5 ml 0.5% agar in culture medium was poured in each well of a 6-well plate. 3000 cells were re-suspended in 1.5 ml top solution consisting of 0.35% agar in culture medium and subsequently seeded on top of the polymerized base solution. All solutions were kept at 40C before pouring to prevent early agar polymerization and to ensure survival of the cells. Plates were incubated at 37C under standard culture conditions for 28--32 days and colony formation was visualized by staining with 0.01% crystal violet and quantified by counting. All experiments were performed in triplicate and experiments were repeated five times with similar results.

Xenografts

Six to eight week old female CD-1 (ICR nu/nu) mice (Charles River Lab-oratories, Sulzberg, Germany) were used and kept under specific pathogen-free conditions according to the guidelines of the Veterinary Office of the Kanton Zurich, Switzerland. Ten million cells re-suspended in sterile PBS were injected subcutaneously on the flanks. Tumor growth was assessed by measuring tumor size once per week.

Immunohistochemistry

Formalin-fixed and paraffin-embedded tumor samples were subjected to immunohistochemical staining for PAX5 and Vimentin as described elsewhere (27). The primary anti-Pax-5 (Transduction Laboratories, Lexington, KY) and anti-Vimentin (DAKO, Glostrup, Denmark) were used.

Transfection with small interfering RNA (siRNA) and measurement of cell proliferation

Briefly, for each experiment 5  105cells/well were plated in 6-well plates and transfected with 80 nM siRNA or scrambled control RNA (scRNA) for 24 h using Lipofectamine 2000 as a transfection mediator according to the manu-facturer's instructions (Invitrogen, Basel, Switzerland). The sequences of the sense strand and of the complementary strand of the siRNA and scRNA were as follows: siRNA sense strand 50-CGGCCACUCGCUUCCGGGCTT-30, complementary strand 50-GCCCGGAAGCGAGUGGCCGTT-30 and scRNA sense strand 50-GCUCCGUCGACUGCGCGCC-30, complementary strand 50-GGCGCGCAGUCGACGGAGCTT-30. These compounds were ordered as double-stranded RNA with a 2-nt DNA overhang (i.e. TT) at both 30-ends (Proligo, Paris, France).

Cell proliferation was measured by use of a colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay as described previously (28). Twenty-four hours after treatment with siRNA or scRNA, cells were plated in 96-well plates and incubated in standard culture medium containing only 5% FCS under standard conditions. The amount of plated cells per well was as follows: LAN-1 and OH1 (1.5  104), Imr32 and MMLI157 (2  104). The first MTT assay was performed immediately after distributing the cells in order to confirm the accuracy of the plating. Cell proliferation was assessed 24, 48, 72 and 96 h after plating. Data recording and analysis was performed as described previously (28). An individual standard calibration curve was constructed for each cell line and absorption values were equalized with the cell number by use of these standard curves.

Results

Expression of PAX5 in N-type neuroblastoma and small cell

lung cancer cell lines

Based on the observation that deregulated expression of

PAX genes is often observed in tumors derived from tissues

in which the respective PAX gene is expressed during normal

organogenesis, we screened tumors of the nervous system for

PAX5 expression by RT--PCR. Expression of PAX5 mRNA

was detected in seven N-type and parental neuroblastoma

cell lines while no transcripts were present in all four S-type

(3)

neuroblastoma cell lines and in two out of three I-type cell

lines (Figure 1A). In order to compare the relative abundance

of the two PAX5 isoforms, which differ only in the first exon

(29), we performed a quantitative Real-Time PCR with primers

specific for isoforms 1A and 1B. All neuroblastoma cell lines

that were positive for PAX5 in the conventional RT--PCR

expressed isoform 1B. It was only in a few cases that a very

low expression of the 1A isoform could be detected as well

(unpublished data). These findings are consistent with data

showing that the 1B promoter is predominantly used for PAX5

expression in non-B lymphocytes (30). PAX5 expression in

N-type and absence in S-type cells could be confirmed at the

protein level by immunoblotting. A major band of the expected

size of ~50 kDa was detected in lysates prepared from N-type

cell lines and the B-cell line Kis-1, but not in S-type cells. The

minor bands above the band representing PAX5 are non-specific

and also visible in S-type cells. An actin control is included

to ensure equal loading (Figure 1B).

As neuroblastoma is a neural crest-derived tumor, we also

screened additional tumors of the neural crest for PAX5 mRNA

expression. While no expression could be detected in five

melanoma cell lines (unpublished data), expression of PAX5

was found in five small cell lung cancer (SCLC) cell lines but

not in five non-small cell lung cancer (NSCLC) cell lines,

which are of endodermal origin (Figure 1C).

In summary, PAX5 expression was detected at the mRNA

and protein level in N-type neuroblastoma and SCLC cell lines

but not in any of the S-type neuroblastoma, melanoma and

non-neural crest-derived NSCLC cell lines, suggesting a role

for PAX5 in the more aggressive phenotype of these cell lines.

Stable expression of PAX5 in S-type neuroblastoma cell line

CA-2E

To investigate the possibility that the differential expression of

PAX5 in N- but not S-type neuroblastoma cell lines could in part

account for their distinctive phenotypes, we generated several

independent clones of the S-type neuroblastoma cell line CA-2E

with stable expression of PAX5. To this end we used the

PAX5-cDNA expression vector pX-13 and the empty cloning vector

pcDNA3.1/Hygro(‡) as a control. A number of clones

trans-fected with either vector pX-13 or the control vector were

isolated after transfection and limiting dilution. Chromosomal

integration of the PAX5 expression vector or the empty cloning

vector was confirmed by vector-specific primers. PAX5

expres-sion in these clones was determined by immunoblotting

(Figure 2A). Three PAX5 expressing clones (P-16, P-25 and

P-28) and three clones transfected with the empty vector (C-43,

C-45 and C-50) were randomly selected for further phenotypic

analysis. The use of a control vector transfected in parallel and

the analysis of three independent clones of each transfected

vector should eliminate the possibility of selecting pre-existing

clones. To exclude this possibility using an additional method,

we performed fluorescence in situ hybridization (FISH)

experi-ments with a MYCN probe (Vysis, Downers Grove, IL). This

experiment showed the presence of two major populations with

either three or four MYCN copies per cell in the untransfected

CA-2E cell line. There was no significant difference in the

number of cells with three or four copies between the P and

C clones, thereby excluding selection of a subclone with a

particular MYCN copy number (unpublished data).

PAX5 expression in CA-2E neuroblastoma cells changes cell

morphology and increases growth rate

Direct microscopy of cell cultures of PAX5 expressing CA-2E

cells revealed distinct morphologic differences compared with

control cells. The latter consisted of flat, epithelial-like cells

with a large cytoplasm with no significant difference to

untransfected CA-2E cells (Figure 2B). In contrast, PAX5

expressing cells became smaller, rounder and were less

attached to plastic which was closer in morphology to N-type

cells (Figure 2C). The observed cell rounding was not due

to apoptosis as shown by propidium iodine staining followed

by FACS analysis (unpublished data).

Growth kinetics were analyzed by seeding 5000 cells of each

of two positive and negative PAX5 clones and counting cell

numbers after different time points. Compared with PAX5

negative clones, the positive clones grew significantly faster

Fig. 1. Expression of PAX5 in neuroblastoma and lung cancer cell lines. Qualitative analysis of PAX5 (upper panels) and GAPDH (lower panels) expression using RT--PCR with cDNA produced as described. (A) S-type neuroblastoma cell ines SH-EP, CA-2E, SH-310 and WSN (lanes 1--4), I-type cell lines SH-IN, ACN and BE(2)C (lanes 5--7), N-type and parental cell lines LAN-1, SH-SY5Y, Imr32, Kelly, BE(2)M17, LAN-5, SK-N-BE(2) (lanes 8--14) and the B cell lymphoma cell line Kis-1 without (lane 15) and with reverse transcriptase (lane 16) as a control. (B) Immunoblot analysis of PAX5 expression using 100 mg of total lysate per lane, for the Kis-1 cell line only 60 mg were loaded. The major band of ~50 kDa represents PAX5 (4), minor bands are non-specific. The actin control is also shown. N-type neuroblastoma cell lines LAN-1, Imr32, LAN-5, SH-SY5Y and SK-N-BE(2) (lanes 1--5) and S-type cell lines SH-EP and CA-2E (lanes 6--7). (C) RT--PCR with cDNA from the SCLC cell lines U1285, NCI-H69, SW2, OH1 and OH3 (lanes 1--5) and the NSCLC cell lines A549, NCI-H125, Calu1, Calu3 and Calu6 (lanes 6--10).

(4)

and to a higher cell number before the growth curves reached

their plateau (Figure 3). Moreover, there was no difference in

growth kinetics between the CA-2E cell line and the control

vector transfected cell line (unpublished data).

PAX5 expression greatly increases anchorage-independent

growth of CA-2E cells

Previous studies have shown that S-type cells isolated from

mixed neuroblastoma cell populations are less tumorigenic

than N-type cells, based on their differential ability for colony

formation in soft agar assays or tumor formation in nude mice

(8). Consistent with these observations CA-2E cells formed

nearly no colonies in our soft agar system, in contrast to the

N-type cell lines SH-SY5Y and LAN-1 (unpublished data).

PAX5 expressing CA-2E clones formed a large number of

colonies that were nevertheless smaller in size compared

with colonies formed by the N-type cell lines SH-SY5Y and

LAN-1. The control clones were comparable with the

wild-type CA-2E cells with almost no colony formation (Figure 4).

These observations could be reproduced for all clones in

several different experiments. Taken together these results

indicate that PAX5 expression can increase

anchorage-independent colony formation when over-expressed in the

appropriate cellular background.

PAX5 expression promotes tumor formation in nude mice

In in vivo experiments the PAX5 expressing P-25 and the

non-expressing C-43 cells were injected into four CD-1 nude mice

each on the lateral flank. Two mice injected with P-25

devel-oped small tumors with a size of 130 and 230 mm

3

after

4 months whereas mice injected with C-43 did not form any

tumors. To confirm this result in a larger population we

injected P-25 cells into seven mice. Although we injected the

cells in both flanks only two mice developed tumors with a size

of 85 mm

3

. Immunohistochemical analysis of the tumors

from both experiments confirmed that they were derived

from P-25 as they expressed PAX5 as well as the CA-2E

marker Vimentin (Figure 5A and B). Immunoreactivity for

PAX5 was present in the nucleus and co-localized with

Vimentin staining in the same cells.

PAX5 down-regulation with siRNA decreases cell

proliferation of N-type neuroblastoma cells

As shown in our growth experiments, stable PAX5 expression

significantly increased the growth rate of CA-2E cells. As

N-type cells already express endogenous PAX5 we exploited

the RNA interference (RNAi) technique (31) in order to

spe-cifically down-regulate PAX5 mRNA. We designed an siRNA

against PAX5 mRNA and an scRNA consisting of the same

Fig. 2. PAX5 expression induces morphological changes in CA-2E cells. The S-type CA-2E neuroblastoma cell line was transfected with the human PAX5 encoding expression vector pX-13 or the empty vector pcDNA3.1/ Hygro(‡) as a control. Independent clones were isolated by limiting dilution and analyzed for PAX5 expression and morphology. (A) Immunoblot of 80 mg total cell lysates obtained from the three PAX5 expressing clones P-16, P-25 and P-28 (lanes 2, 4 and 6) and from three control clones C-43, C-45 and C-50 (lanes 1, 3 and 5) (upper panel). The equal amount of lysate was verified by a specific staining for actin (lower panel). Photomicrographs of cell cultures from the representative PAX5 expressing clone P-28 (C) and from the control clone C-45 (B) are also shown and a 50 mm scale is indicated. PAX5 expressing cells are smaller, rounder and less attached to the culture flask.

Fig. 3. Increased proliferation of PAX5 expressing CA-2E cells. 5000 cells of two PAX5 expressing (P-16 and P-25) and two non-expressing clones (C-43 and C-45) were plated and growth was recorded. Cell numbers were assessed with a Coulter counter at indicated time points. Cells were first counted on day 5, which is set as zero in the figure. The experiment was performed in triplicate for each clone and time point. Means and standard deviations are shown.

(5)

bases organized in a randomized combination without any

homology to human genomic sequences. The N-type

neuro-blastoma cell lines LAN-1 and Imr32 as well as the SCLC cell

line OH1 were either transfected with siRNA or the same

amount of scRNA. The melanoma cell line MMLI157, which

does not express endogenous PAX5 was used as a control.

Immunoblot analysis of cells transfected with siRNA and

har-vested 72 h after transfection showed a down-regulation of

the PAX5 protein level by 32% in LAN-1 cells and by 12% in

Imr32 and OH1 cells when compared with scRNA treatment

(Figure 6A--C). Staining for actin was used as a loading control

and is also shown.

Proliferation of transfected cells was measured by a MTT

assay. A modest but significant decrease in growth of the

siRNA transfected cells compared with scRNA transfected

cells was first observed 48 h after plating (Figure 6A--C).

The effect was greatest in LAN-1 where the PAX5

down-regulation worked most effective and small in OH1 where

PAC5 was weakly down-regulated. In addition the control

cell line MMLI157 was unaffected by either treatment (Figure

6D).

Discussion

In this study we report the expression of the oncogenic

transcription factor PAX5 in a subset of neuroblastoma cell

lines characterized by an aggressive phenotype, the so-called

`N-type' cells. In contrast, PAX5 was not expressed in any

of the tested S-type neuroblastoma cell lines. Further screening

of neural crest-derived tumors demonstrated expression in

SCLC but not in melanoma cell lines. The data show a distinct

expression pattern of PAX5 in neural crest-derived tumors with

a neuronal or neuroendocrine differentiation, indicating that

PAX5 might be a valuable marker for these derivatives. The

lack of expression in both melanoma cell lines and S-type cell

lines is consistent with the model of S-type cells being

differ-entiated along the melanocyte pathway (4).

More significantly, the expression of PAX5 in N-type

neuro-blastoma cells might be an important determinant of the more

malignant phenotype of these cells. To test this hypothesis two

approaches were taken. First, PAX5 was over-expressed in the

S-type neuroblastoma cell line CA-2E, resulting in a

remark-able phenotypic change. Secondly, PAX5 was down-regulated

in N-type cell lines and SCLC cell line by means of the siRNA

technique. A recent report has shown the coexistence of

subpopulations with different MYCN copy numbers in the

neuroblastoma cell line SK-N-SH and the derived cell lines

SH-SY5Y (N-type) and SH-EP (S-type). It was suggested that

the phenomenon of phenotypic interconversion is likely to

result from selection of pre-existing clones that can outgrow

Fig. 4. PAX5 expressing CA-2E cells are capable of growing anchorage-independently. The soft agar assay was performed with three PAX5 expressing (P-16, P-25 and P-28) and three PAX5 non-expressing clones (C-43, C-45 and C-50) derived from the CA-2E neuroblastoma cell line. (A) The data summarized in the table represent the means and standard deviations of a representative experiment carried out in triplicate. The lower panel shows pictures of the PAX5 expressing clone P-16 (B) and of the

non-expressing clone C-43 (C) after staining with 0.01% crystal violet. Fig. 5. Tumors established from the CA-2E transfected clone P-25 express markers PAX5 and Vimentin. Paraffin sections of a xenograft of the PAX5 expressing CA-2E clone P-25 established in a CD-1 nude mouse are shown. The tumor was isolated 4 months after injection into the lateral flank with a size of 230 mm3. Immunohistochemical stainings for PAX5 (A) and Vimentin (B) were performed as described. Magnification is 280-fold.

(6)

under certain conditions (32). The selection of a subclone with

a higher MYCN copy number could therefore account for the

increase in aggressiveness seen in our clones transfected with

PAX5. To exclude this important possibility we performed

FISH analysis with the MYCN probe in the untransfected

CA-2E cell line and the transfected clones. We could indeed

detect the coexistence of two populations with respect to

MYCN copy number, but no evidence of selection after

trans-fection. Therefore, these results provide evidence that the

phenotypic changes observed by us, including the increase of

tumorigenicity, are a result of the expression of PAX5 in

CA-2E cells.

The cellular background of the CA-2E cell line proved to be

particularly permissive to mediate the effects of PAX5

expres-sion, most likely because interacting co-factors required for

PAX5 function are present. CA-2E was established from the

bone marrow aspirate of a 16-month-old patient with

pro-gressive disease (33). The cells have the typical morphology

of S-type cells and do not form colonies in a soft agar assay.

PAX5 expression resulted in a striking increase in colony

numbers. The size of the individual colonies, however, was

smaller than those formed by the N-type cell line SH-SY5Y.

Consistent with this observation, PAX5 expressing cells

formed tumors only with low efficiency in animals. These

results are reminiscent of observations made when analysing

the transforming capacity of the PAX3--FKHR fusion protein

(34). Expression of this fusion protein in chicken embryo

fibroblasts led to cellular transformation in vitro, as shown

in focus and soft agar colony assays. In animal experiments,

however, no in vivo transforming capacity could be

demon-strated. It is possible that tumor induction in vivo requires

additional genetic changes or induction of additional genes

(e.g. responsible for neo-angiogenesis) that are not required

for colony formation in soft agar. In addition, several other

attempts to induce tumors in transgenic models by

over-expressing PAX5 or PAX3-FKHR have failed (35--37). These

Fig. 6. Down-regulation of PAX5 expression in N-type neuroblastoma and SCLC cell lines leads to a reduced growth rate. Endogenous PAX5 expression in the N-type neuroblastoma cell lines LAN-1 (A) and Imr32 (B) as well as in the SCLC cell line OH1 (C) was targeted by transfection of 80 nM of a sequence specific siRNA or 80 nM of a scrambled control scRNA. PAX5 protein level was determined 72 h after transfection by an immunoblot using 100 mg from total lysates (A--C). Loading of equal amounts was verified by immunoblotting for actin. The proliferation of transfected cells was assessed using the MTT colorimetric assay at indicated time points (A--C). MMLI157 was thereby used as a negative control (D). The number of cells was calculated according to a calibration curve to correlate optical density and cell number. An individual standard curve was constructed for each cell line. The data represent mean values and standard deviations of an experiment carried out in quintuplicate.

(7)

results could indicate that deregulated expression of PAX

genes is insufficient to cause tumor formation in vivo.

However, these results do not exclude an important role for

PAX gene expression in tumor progression or maintenance.

Our observation that targeting of PAX5 expression in N-type

neuroblastoma cell lines decreases proliferation supports this

hypothesis.

Our results confirm other observations implicating mouse

Pax5 as a positive regulator of cell proliferation (38). In

addi-tion down-regulaaddi-tion of PAX5 in N-type cells affected

growth, and we are currently trying to identify the target genes

mediating this effect. A novel finding is our observation that

PAX5 expression had a strong effect on S-type cell morphology

and adhesion. This suggests that PAX5 might also regulate

expression of cytoskeletal components and/or proteins of

extracellular matrix.

Our study is limited by the fact that the effect of PAX5

over-expression was studied in only one S-type cell line.

Never-theless, the expression data and the effects of PAX5 expression

in the S-type neuroblastoma cell line CA-2E suggest a role for

PAX5 in the molecular pathogenesis of neuroblastoma tumors.

Whether PAX5 might serve as a molecular target for therapy

is not known. To investigate this possibility we have started

an analysis of PAX5 expression in tumor samples. Recent

findings of widespread expression of PAX genes in numerous

common tumors and dependence of tumor viability on that

expression suggest that PAX5 and other PAX transcription

factors might be valuable targets for cancer therapy (39). It is

clear however, that the PAX5 transfected CA-2E cell line is

a very useful model system to study the oncogenic function of

PAX5. For example, the target genes mediating the oncogenic

effect of PAX5 can be identified using microarray technology.

Using this technique we have already identified several target

genes that are regulated by PAX5 also in cell lines other than

neuroblastoma cells (manuscript in preparation). The results

of these studies will provide answers on how PAX5 performs

its oncogenic function.

Acknowledgements

The authors are grateful to Thomas Stallmach who performed numerous immunohistochemical stainings of our mice tumor sections. We thank Sally Donaldson for her valuable scientific input and the critical reading of the manuscript and we thank Dario Neri for supporting this research project. This study was funded by a grant of the Swiss Cancer League to A.H.

References

1.Brodeur,G.M. (2003) Neuroblastoma: biological insights into a clinical enigma. Nature Rev. Cancer, 3, 203--216.

2.Spix,C., Aareleid,T., Stiller,C., Magnani,C., Kaatsch,P. and Michaelis,J. (2001) Survival of children with neuroblastoma. Time trends and regional differences in Europe, 1978--1992. Eur. J. Cancer, 37, 722--729. 3.Le Douarin,N. and Kalcheim,C. (1999) The Neural Crest. Cambridge

University Press, Cambridge.

4.Ciccarone,V., Spengler,B.A., Meyers,M.B., Biedler,J.L. and Ross,R.A. (1989) Phenotypic diversification in human neuroblastoma cells: expres-sion of distinct neural crest lineages. Cancer Res., 49, 219--225. 5.Thiele,C.J. (1999) Neuroblastoma. In Masters,J.R.W. and Palsson,B. (eds),

Human Cell Culture. Kluwer Academic Publishers, Dordrecht, The Netherlands, Vol. 1, pp. 21--53.

6.Ross,R.A., Spengler,B.A., Domenech,C., Porubcin,M., Rettig,W.J. and Biedler,J.L. (1995) Human neuroblastoma I-type cells are malignant neural crest stem cells. Cell Growth Differ., 6, 449--456.

7.Ross,R.A., Hein,A.M., Braca,J.A.,3rd, Spengler,B.A., Biedler,J.L. and Scammell,J.G. (2002) Glucocorticoids induce neuroendocrine cell

differentiation and increase expression of N-myc in N-type human neuroblastoma cells. Oncol. Res., 13, 87--94.

8.Biedler,J.L., Spengler,B.A.,T.,C. and Ross,R.A. (1988) Transdifferentiation of human neuroblastoma cells results in coordinate loss of neuronal and malignant properties. In Evans,A., D'Angio,G., Knudson,A. and Seeger,R. (eds), Advances in Neuroblastoma Research 2. Alan R. Liss, Inc., New York, pp. 265--276.

9.La Quaglia,M.P. and Manchester,K.M. (1996) A comparative analysis of neuroblastic and substrate-adherent human neuroblastoma cell lines. J. Pediatr. Surg., 31, 315--318.

10.Isaacs,J.S., Hardman,R., Carman,T.A., Barrett,J.C. and Weissman,B.E. (1998) Differential subcellular p53 localization and function in N- and S-type neuroblastoma cell lines. Cell Growth Differ., 9, 545--555. 11.Sadee,W.,Yu,V.C., Richards,M.L., Preis,P.N., Schwab,M.R., Brodsky,F.M.

and Biedler,J.L. (1987) Expression of neurotransmitter receptors and myc protooncogenes in subclones of a human neuroblastoma cell line. Cancer Res., 47, 5207--5212.

12.Foley,J., Cohn,S.L., Salwen,H.R., Chagnovich,D., Cowan,J., Mason,K.L. and Parysek,L.M. (1991) Differential expression of N-myc in pheno-typically distinct subclones of a human neuroblastoma cell line. Cancer Res., 51, 6338--6345.

13.Hopkins-Donaldson,S., Bodmer,J.L., Bourloud,K.B., Brognara,C.B., Tschopp,J. and Gross,N. (2000) Loss of caspase-8 expression in highly malignant human neuroblastoma cells correlates with resistance to tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis. Cancer Res., 60, 4315--4319.

14.Eggert,A., Grotzer,M.A., Zuzak,T.J., Wiewrodt,B.R., Ikegaki,N. and Brodeur,G.M. (2000) Resistance to TRAIL-induced apoptosis in neuro-blastoma cells correlates with a loss of caspase-8 expression. Med. Pediatr. Oncol., 35, 603--607.

15.Chi,N. and Epstein,J.A. (2002) Getting your Pax straight: Pax proteins in development and disease. Trends Genet., 18, 41--47.

16.Asano,M. and Gruss,P. (1992) Pax-5 is expressed at the midbrain-hindbrain boundary during mouse development. Mech. Dev., 39, 29--39. 17.Gerard,M., Abitbol,M., Delezoide,A.L., Dufier,J.L., Mallet,J. and

Vekemans,M. (1995) PAX-genes expression during human embryonic development, a preliminary report. C. R. Acad. Sci. III, 318, 57--66. 18.Adams,B., Dorfler,P., Aguzzi,A., Kozmik,Z., Urbanek,P., Maurer-Fogy,I.

and Busslinger,M. (1992) Pax-5 encodes the transcription factor BSAP and is expressed in B lymphocytes, the developing CNS and adult testis. Genes Dev., 6, 1589--1607.

19.Urbanek,P., Fetka,I., Meisler,M.H. and Busslinger,M. (1997) Cooperation of Pax2 and Pax5 in midbrain and cerebellum development. Proc. Natl Acad. Sci USA, 94, 5703--5708.

20.Schafer,B.W. (1998) Emerging roles for PAX transcription factors in cancer biology. Gen. Physiol. Biophys., 17, 211--224.

21.Kozmik,Z., Sure,U., Ruedi,D., Busslinger,M. and Aguzzi,A. (1995) Deregulated expression of PAX5 in medulloblastoma. Proc. Natl Acad. Sci. USA, 92, 5709--5713.

22.Krenacs,L., Himmelmann,A.W., Quintanilla-Martinez,L., Fest,T., Riva,A., Wellmann,A., Bagdi,E., Kehrl,J.H., Jaffe,E.S. and Raffeld,M. (1998) Transcription factor B-cell-specific activator protein (BSAP) is differen-tially expressed in B cells and in subsets of B-cell lymphomas. Blood, 92, 1308--1316.

23.Scholl,F.A., Kamarashev,J., Murmann,O.V., Geertsen,R., Dummer,R. and Schafer,B.W. (2001) PAX3 is expressed in human melanomas and contributes to tumor cell survival. Cancer Res., 61, 823--826.

24.Hopkins-Donaldson,S., Ziegler,A., Kurtz,S., Bigosch,C., Kandioler,D., Ludwig,C., Zangemeister-Wittke,U. and Stahel,R. (2003) Silencing of death receptor and caspase-8 expression in small cell lung carcinoma cell lines and tumors by DNA methylation. Cell Death Differ., 10, 356--364. 25.Adshead,J.M., Ogden,C.W., Penny,M.A., Stuart,E.T. and Kessling,A.M.

(1999) The expression of PAX5 in human transitional cell carcinoma of the bladder: relationship with de-differentiation. Br. J. Urol. Int., 83, 1039--1044.

26.Sambrook,J. and Russell,D.W. (2001) Molecular Cloning A Laboratory Manual. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York.

27.Koesters,R., Niggli,F., von Knebel Doeberitz,M. and Stallmach,T. (2003) Nuclear accumulation of beta-catenin protein in Wilms' tumours. J. Pathol., 199, 68--76.

28.Olie,R.A., Hall,J., Natt,F., Stahel,R.A. and Zangemeister-Wittke,U. (2002) Analysis of ribosyl-modified, mixed backbone analogs of a bcl-2/bcl-xL antisense oligonucleotide. Biochim. Biophys. Acta, 1576, 101--109. 29.Busslinger,M., Klix,N., Pfeffer,P., Graninger,P.G. and Kozmik,Z. (1996)

(8)

locus adjacent to two alternative PAX-5 promoters in a diffuse large-cell lymphoma. Proc. Natl Acad. Sci. USA, 93, 6129--6134.

30.Morrison,A.M., Jager,U., Chott,A., Schebesta,M., Haas,O.A. and Busslinger,M. (1998) Deregulated PAX-5 transcription from a translo-cated IgH promoter in marginal zone lymphoma. Blood, 92, 3865--3878. 31.Fire,A., Xu,S., Montgomery,M.K., Kostas,S.A., Driver,S.E. and Mello,C.C.

(1998) Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature, 391, 806--811.

32.Cohen,N., Betts,D.R., Rechavi,G., Amariglio,N. and Trakhtenbrot,L. (2003) Clonal expansion and not cell interconversion is the basis for the neuroblast and nonneuronal types of the SK-N-SH neuroblastoma cell line. Cancer Genet. Cytogenet., 143, 80--84.

33.Suardet,L., Gross,N., Gaide,A.C., Beck,D. and Eliason,J.F. (1989) Epidermal growth factor responsiveness of a new human neuroblastoma cell line. Int. J. Cancer, 44, 661--668.

34.Scheidler,S., Fredericks,W.J., Rauscher,F.J.,3rd, Barr,F.G. and Vogt,P.K. (1996) The hybrid PAX3-FKHR fusion protein of alveolar rhabdomyo-sarcoma transforms fibroblasts in culture. Proc. Natl Acad. Sci. USA, 93, 9805--9809.

35.Anderson,M.J., Shelton,G.D., Cavenee,W.K. and Arden,K.C. (2001) Embryonic expression of the tumor-associated PAX3-FKHR fusion pro-tein interferes with the developmental functions of Pax3. Proc. Natl Acad. Sci. USA, 98, 1589--1594.

36.Lagutina,I., Conway,S.J., Sublett,J. and Grosveld,G.C. (2002) Pax3-FKHR knock-in mice show developmental aberrations but do not develop tumors. Mol. Cell. Biol., 22, 7204--7216.

37.Steinbach,J.P., Kozmik,Z., Pfeffer,P. and Aguzzi,A. (2001) Overexpression of Pax5 is not sufficient for neoplastic transformation of mouse neuroectoderm. Int. J. Cancer, 93, 459--467.

38.Wakatsuki,Y., Neurath,M.F., Max,E.E. and Strober,W. (1994) The B cell-specific transcription factor BSAP regulates B cell proliferation. J. Exp. Med., 179, 1099--1108.

39.Muratovska,A., Zhou,C., He,S., Goodyer,P. and Eccles,M.R. (2003) Paired-box genes are frequently expressed in cancer and often required for cancer cell survival. Oncogene, 22, 6045--6053.

Figure

Fig. 1. Expression of PAX5 in neuroblastoma and lung cancer cell lines.
Fig. 3. Increased proliferation of PAX5 expressing CA-2E cells. 5000 cells of two PAX5 expressing (P-16 and P-25) and two non-expressing clones (C-43 and C-45) were plated and growth was recorded
Fig. 4. PAX5 expressing CA-2E cells are capable of growing anchorage- anchorage-independently
Fig. 6. Down-regulation of PAX5 expression in N-type neuroblastoma and SCLC cell lines leads to a reduced growth rate

Références

Documents relatifs

It lies – first – in current historical work on ancient mathematics carried out, in particular, in the SPHERE 13 research group; second, in what we feel was a fruitful introduction,

Supplementary Figure 2: Cell cycle analysis of neuroblastoma cell lines treated with 20nM camptothecin (CPT) or topotecan (TPT) and harvested at different time points, as

To filter the hypomethylated genes for those likely to be functionally important in neuroblastoma pathogenesis, we used publicly available databases to search for genes that; (1)

Quinoxalines 3a–b, 4a–b, 7, 8, 9a–b, 10a–b, 15, 16 and clinical compound XK469 were evaluated for their in vitro anti-proliferative activity on a panel of SK-N-SH and IMR-32

The self-exciting Faraday-disk homopolar dynamo similar to the differentially rotating conducting cylinder with helical surface waves on its top.. be negligibly small, as they

Supplementary Materials: The following are available online at http://www.mdpi.com/2072-6694/11/2/202/s1 , Table S1: Clinical features of neuroblastoma tumors, Table S2: Cell

Additional file 9 List of genes with rare non-synonymous SNVs, indels, substitutions, copy number variations or structural variations in SH-SY5Y and lists of genes used to

The 5' boundary of SLC11A1 predicted upstream enhancer element is indicated by moderately intense cell-type specific histone marks H3K4me1 and H3K27ac, which correspond to the